Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Publication year range
1.
Transfusion ; 64(5): 906-918, 2024 May.
Article in English | MEDLINE | ID: mdl-38530740

ABSTRACT

BACKGROUND: To identify specific human neutrophil antigen (HNA) antibodies, assays using neutrophils such as monoclonal antibody-specific immobilization of granulocyte antigens (MAIGA) are recommended. However, these assays are limited by labor-intensive neutrophil preparation and varying antigen expression levels. METHODS: We evaluated a newly developed immunocomplex capture fluorescence assay (ICFA) for identifying HNA-1 antibodies and compared it to MAIGA and LABScreen Multi (LABM), which utilizes recombinant HNA-coated Luminex beads. For ICFA, HNA-1a or HNA-1b transfected cells replaced neutrophils. Cells incubated with serum were lysed, and immune complexes were captured using five CD16 monoclonal antibody-conjugated Luminex beads. Nine antisera with known specificity and 26 samples suspected of containing HNA antibodies were analyzed by ICFA and MAIGA using neutrophils or transfected cells (ICFA-N or ICFA-T, and MAIGA-N or MAIGA-T, respectively). RESULTS: ICFA-T and MAIGA-N accurately determined the specificity of all antibodies in the nine antiserum samples. The ICFA-T detection limit was 2048-fold for anti-HNA-1a and 256-fold for anti-HNA-1b; the limits of MAIGA-T, MAIGA-N, and LABM were 32-, 4 ~ 64-, and 128-fold for anti-HNA-1a and 64-, 16 ~ 64-, and 32-fold for anti-HNA-1b, respectively. Twelve and 7 of the remaining 26 samples tested negative and positive, respectively, in both ICFA-T and MAIGA-N. Antibody specificity against HNA-1a or HNA-1b determined using ICFA-T agreed with that determined using MAIGA-N and LABM. Another seven samples tested positive in ICFA-T but negative in MAIGA-N. CONCLUSION: The novel ICFA is highly sensitive and exhibits specificity similar to MAIGA and LABM for detecting HNA-1 antibodies.


Subject(s)
Immunoassay , Isoantigens , Neutrophils , Humans , Antibodies, Monoclonal/immunology , Isoantigens/chemistry , Isoantigens/immunology , Neutrophils/immunology , Transfection , Fluorescent Dyes/chemistry , Immunoassay/methods
2.
Transfusion ; 60(4): 815-821, 2020 04.
Article in English | MEDLINE | ID: mdl-32072650

ABSTRACT

BACKGROUND: Neutrophil specific Fcγ receptor IIIb (CD16b) is a low-affinity IgG receptor. Its polymorphic variants are associated with human neutrophil antigens (HNA). HNA-1a and HNA-1b differ in four amino acids. Immunization can lead to the production of alloantibodies. The exact contribution of four amino acid exchanges for the formation of HNA-1a, -1b epitopes is currently unknown. STUDY DESIGN AND METHODS: Permutation of each polymorphic amino acid from wild-type CD16b cDNA constructs was performed and expressed on HEK293 cells. All 16 receptor variants were produced and tested against 19 well-characterized HNA antisera in an antigen capture assay. RESULTS: Analyzing the reaction pattern revealed that anti-HNA-1a antibodies can bind whenever asparagine (N) is present in position 65, regardless of the three other positions (CD16b *N**). Anti-HNA-1b antibodies can bind when serine (S) is present in position 36 (CD16b S***), when N is present in position 82 (CD16b **N*), or both (CD16b S*N*). CD16b variants with N65 and S36 and/or N82 (such as CD16b SNN*) bind both, anti-HNA-1a and anti-HNA-1b alloantibodies. If these specific amino acids are missing (as in CD16b RSD*), no antibodies will bind. CONCLUSION: Whereas the primary structure of HNA-1a and HNA-1b usually differs in four amino acids, epitope composition is not "antithetical". N65 alone determines the presence of HNA-1a, and S36 and/or N82 determine the presence of HNA-1b. Amino acid 106 does not participate in epitope formation. Our findings are of specific relevance when a HNA-1 phenotype is predicted from a genotype.


Subject(s)
Isoantigens/immunology , Receptors, IgG/immunology , Amino Acid Sequence , Antigen-Antibody Complex/genetics , Binding Sites, Antibody/genetics , DNA, Complementary , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Genetic Variation , HEK293 Cells , Humans , Isoantibodies/metabolism , Isoantigens/chemistry , Receptors, IgG/genetics
3.
J Immunol ; 201(12): 3780-3792, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30429288

ABSTRACT

In transplantation, development of humoral alloimmunity against donor HLA is a major cause of organ transplant failure, but our ability to assess the immunological risk associated with a potential donor-recipient HLA combination is limited. We hypothesized that the capacity of donor HLA to induce a specific alloantibody response depends on their structural and physicochemical dissimilarity compared with recipient HLA. To test this hypothesis, we first developed a novel computational scoring system that enables quantitative assessment of surface electrostatic potential differences between donor and recipient HLA molecules at the tertiary structure level [three-dimensional electrostatic mismatch score (EMS-3D)]. We then examined humoral alloimmune responses in healthy females subjected to a standardized injection of donor lymphocytes from their male partner. This analysis showed a strong association between the EMS-3D of donor HLA and donor-specific alloantibody development; this relationship was strongest for HLA-DQ alloantigens. In the clinical transplantation setting, the immunogenic potential of HLA-DRB1 and -DQ mismatches expressed on donor kidneys, as assessed by their EMS-3D, was an independent predictor of development of donor-specific alloantibody after graft failure. Collectively, these findings demonstrate the translational potential of our approach to improve immunological risk assessment and to decrease the burden of humoral alloimmunity in organ transplantation.


Subject(s)
Graft Rejection/immunology , HLA-DQ Antigens/chemistry , HLA-DRB1 Chains/chemistry , Immunity, Humoral , Isoantibodies/biosynthesis , Isoantigens/chemistry , Kidney Transplantation , Female , Graft Rejection/diagnosis , HLA-DQ Antigens/immunology , HLA-DRB1 Chains/immunology , Histocompatibility , Histocompatibility Testing , Humans , Isoantigens/immunology , Male , Static Electricity , Tissue Donors , Transplant Recipients
4.
Am J Transplant ; 19(6): 1708-1719, 2019 06.
Article in English | MEDLINE | ID: mdl-30414349

ABSTRACT

Alloimmune risk stratification in renal transplantation has lacked the necessary prognostic biomarkers to personalize recipient care or optimize clinical trials. HLA molecular mismatch improves precision compared to traditional antigen mismatch but has not been studied in detail at the individual molecule level. This study evaluated 664 renal transplant recipients and correlated HLA-DR/DQ single molecule eplet mismatch with serologic, histologic, and clinical outcomes. Compared to traditional HLA-DR/DQ whole antigen mismatch, HLA-DR/DQ single molecule eplet mismatch improved the correlation with de novo donor-specific antibody development (area under the curve 0.54 vs 0.84) and allowed recipients to be stratified into low, intermediate, and high alloimmune risk categories. These risk categories were significantly correlated with primary alloimmune events including Banff ≥1A T cell-mediated rejection (P = .0006), HLA-DR/DQ de novo donor-specific antibody development (P < .0001), antibody-mediated rejection (P < .0001), as well as all-cause graft loss (P = .0012) and each of these correlations persisted in multivariate models. Thus, HLA-DR/DQ single molecule eplet mismatch may represent a precise, reproducible, and widely available prognostic biomarker that can be applied to tailor immunosuppression or design clinical trials based on individual patient risk.


Subject(s)
HLA-DQ Antigens/immunology , HLA-DR Antigens/immunology , Kidney Transplantation , Transplantation Immunology , Adult , Biomarkers/blood , Child , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Female , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/genetics , HLA-DR Antigens/chemistry , HLA-DR Antigens/genetics , Histocompatibility Testing , Humans , Isoantigens/chemistry , Isoantigens/immunology , Kidney Transplantation/adverse effects , Male , Middle Aged , Models, Molecular , Prognosis , Risk Factors , Tissue Donors
5.
Cell Immunol ; 314: 18-25, 2017 04.
Article in English | MEDLINE | ID: mdl-28108005

ABSTRACT

We investigated the regulatory activity of human adipose-derived mesenchymal stromal cells (MSCs) (n=10) towards immune cells in a cohort of 84 rheumatoid arthritis (RA) patients, 36 apparently healthy controls. We co-cultured MSCs with lymphocyte subsets of T, B, and T regulatory cells (Tregs). Levels of the pro- and anti-inflammatory markers (tumor necrosis factor alpha (TNF-α), interferon gamma (IFN-γ), and interleukin-10 (IL-10)) were estimated in serum and co-culture supernatants. The study revealed a two-fold increase in the proportion of Tregs and an increased level of CD4+CD25+FoxP3. MSCs altered T cell, B cell, and Treg cytokine production during an anti-inflammatory immune response. The MSCs inhibited CD3+T cell-mediated TNF-α secretion, upregulated IL-10, and suppressed the production of autoantibodies against citrullinated protein antigens produced by B cells. These data offer insight into the interactions between allogeneic MSCs and immune cells, and elucidate the dose-dependent modulation of MSCs.


Subject(s)
Adipose Tissue/pathology , Arthritis, Rheumatoid/therapy , Immunotherapy/methods , Mesenchymal Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Antibodies/blood , Arthritis, Rheumatoid/immunology , Cells, Cultured , Citrulline/chemistry , Coculture Techniques , Cytokines/metabolism , Female , Humans , Immunity , Inflammation Mediators/metabolism , Isoantigens/chemistry , Isoantigens/immunology , Male , Middle Aged , Young Adult
6.
J Biol Chem ; 286(46): 40163-73, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21937447

ABSTRACT

Modification of the primary anchor positions of antigenic peptides to improve binding to major histocompatibility complex (MHC) proteins is a commonly used strategy for engineering peptide-based vaccine candidates. However, such peptide modifications do not always improve antigenicity, complicating efforts to design effective vaccines for cancer and infectious disease. Here we investigated the MART-1(27-35) tumor antigen, for which anchor modification (replacement of the position two alanine with leucine) dramatically reduces or ablates antigenicity with a wide range of T cell clones despite significantly improving peptide binding to MHC. We found that anchor modification in the MART-1(27-35) antigen enhances the flexibility of both the peptide and the HLA-A*0201 molecule. Although the resulting entropic effects contribute to the improved binding of the peptide to MHC, they also negatively impact T cell receptor binding to the peptide·MHC complex. These results help explain how the "anchor-fixing" strategy fails to improve antigenicity in this case, and more generally, may be relevant for understanding the high specificity characteristic of the T cell repertoire. In addition to impacting vaccine design, modulation of peptide and MHC flexibility through changes to antigenic peptides may present an evolutionary strategy for the escape of pathogens from immune destruction.


Subject(s)
Antigens, Neoplasm/chemistry , HLA-A2 Antigen/chemistry , Isoantigens/chemistry , Peptide Fragments/chemistry , Receptors, Antigen, T-Cell/chemistry , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , HLA-A2 Antigen/immunology , Humans , Isoantigens/immunology , Peptide Fragments/immunology , Protein Binding , Protein Structure, Quaternary , Receptors, Antigen, T-Cell/immunology , Structure-Activity Relationship
7.
Transfusion ; 52(7): 1458-67, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22211383

ABSTRACT

BACKGROUND: HNA-3 is a diallellic system located on choline transporter-like protein 2 (CTL2), defined by a polymorphism at Amino Acid 154. HNA-3a antibodies are of clinical importance in transfusion-related acute lung injury but antibody detection requires labor-intensive granulocyte isolation from HNA-typed donors and the use of techniques such as the granulocyte agglutination test or granulocyte immunofluorescence test. Also, there is no commercial test for detection of HNA-3 antibodies. STUDY DESIGN AND METHODS: HEK293 cells were transfected to generate stable cell lines expressing CTL2 fragments (Amino Acids 55-230) and full-length membrane bound CTL2 with HNA-3a and -3b epitopes. Soluble fragments were used in enzyme-linked immunosorbent assays to detect HNA-3 antibodies. The cell lines expressing full-length proteins were trypsin treated to remove HLA antigens and frozen at -80°C. Thawed cells were then used to detect HNA-3 antibodies by flow cytometry. RESULTS: Glycosylated and soluble CTL2 fragments were correctly recognized by 15 of 31 anti-HNA-3a sera and by both available anti-HNA-3b sera. Twenty-one anti-HLA sera reacted variably with untreated cell lines expressing full-length CTL2. After trypsin treatment of the cell lines, reactivity with HLA antisera was abrogated and all 31 anti-HNA-3a and two anti-HNA-3b sera bound to the corresponding cell line. CONCLUSION: Whereas soluble, glycosylated CTL2 fragments cannot be used for the detection of HNA-3 antibodies, the HEK293 cells expressing full-length CTL2 proteins were useful in the detection of HNA-3 antibodies even in the presence of HLA antibodies. Moreover, the cell lines can be stored for at least 6 months before use.


Subject(s)
Flow Cytometry/methods , Isoantibodies/analysis , Isoantigens/chemistry , Membrane Glycoproteins/chemistry , Membrane Transport Proteins/chemistry , Serum/chemistry , Gene Expression , HEK293 Cells , Humans , Isoantibodies/blood , Isoantibodies/chemistry , Isoantibodies/immunology , Isoantigens/biosynthesis , Isoantigens/genetics , Isoantigens/immunology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Membrane Transport Proteins/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Serum/immunology , Serum/metabolism , Transfection
8.
Transfusion ; 51(10): 2160-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21756260

ABSTRACT

BACKGROUND: Severe transfusion-related acute lung injury is often caused by antibodies directed against the human neutrophil alloantigen (HNA)-3a. HNA-3a results from an amino acid exchange (Arg154Gln) in the first extracellular loop of the choline transporter-like protein 2 (CTL2). The characteristics of the binding domain(s) of HNA-3a antibodies are unknown. STUDY DESIGN AND METHODS: For epitope mapping, a library of 23 different HNA-3a (R(154)) and three HNA-3b (Q(154)) peptides covering different parts of the first extracellular loop of CTL2 (aa(55-231)) was synthesized in Escherichia coli and tested by Western blot with two HNA-3a alloantibody-containing plasma samples and by enzyme immunoassay (EIA) with different HNA-3a- (n = 21) and HNA-3b- (n = 1) positive plasma samples. RESULTS: Despite promising Western blot results using highly reactive plasma samples, we found widely varying reactivities of different HNA-3a plasmas in the EIA, with only 11 of 21 HNA-3a antibodies binding to any of the tested HNA-3a peptides and with no peptide recognized by more than nine of the 21 antibodies. The HNA-3b plasma did not react with R(154) peptides in the EIA nor with R(154) or Q(154) peptides in Western blot experiments. Plasma reactivity profiles with the peptides did not correlate with those observed using granulocyte agglutination and granulocyte immunofluorescence tests. CONCLUSION: Binding of HNA-3a alloantibodies depends on the conformation of the intact CTL2 protein and their binding sites may differ substantially. Peptide-based assays for detection of HNA-3a antibodies bear the risk to be insensitive and require systematic validation with a large panel of antibodies.


Subject(s)
Epitope Mapping , Isoantibodies/immunology , Isoantigens/immunology , Plasma/immunology , Blood Transfusion , Blotting, Western , Female , Humans , Immunoenzyme Techniques , Isoantibodies/chemistry , Isoantigens/chemistry , Membrane Glycoproteins/chemistry , Membrane Transport Proteins/chemistry , Middle Aged
9.
Proteins ; 78(11): 2506-22, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20602356

ABSTRACT

The multi-domain protein von Willebrand factor is crucial in the blood coagulation process at high shear. The A1 domain binds to the platelet surface receptor glycoprotein Ibalpha (GpIb alpha) and this interaction is known to be strengthened by tensile force. The molecular mechanism behind this observation was investigated here by molecular dynamics simulations. The results suggest that the proteins unbind through two distinct pathways depending whether a high-tensile force is applied or whether unbinding happens through thermal fluctuations. In the high-force unbinding pathway the A1 domain was observed to rotate away from the C-terminus of GpIb alpha. In contrast, during thermal unbinding the A1 domain rotated in the opposite direction as in the high-force pathway and the distance between the terminii of A1 and the GpIb alpha C-terminus shortened. This shortening was reduced and the lifetime of the bond extended if a moderate tensile force was applied across the complex. This suggests that the thermal unbinding pathway is inhibited by a moderate tensile force which is in agreement with the catch bond property shown previously in single molecule experiments. A designed mutant of GpIb alpha is suggested here in order to test in vitro the thermal unbinding pathway observed in silico.


Subject(s)
Isoantigens/chemistry , Molecular Dynamics Simulation , von Willebrand Factor/chemistry , Biomechanical Phenomena , Hot Temperature , Hydrogen Bonding , Isoantigens/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Mutation , Platelet Glycoprotein GPIb-IX Complex , Principal Component Analysis , Protein Binding , Protein Conformation , von Willebrand Factor/metabolism
10.
J Exp Med ; 195(9): 1175-86, 2002 May 06.
Article in English | MEDLINE | ID: mdl-11994422

ABSTRACT

The crystal structures of the 2C/H-2K(bm3)-dEV8 allogeneic complex at 2.4 A and H-2K(bm3)-dEV8 at 2.15 A, when compared with their syngeneic counterparts, elucidate structural changes that induce an alloresponse. The Asp77Ser mutation that imbues H-2K(bm3)-dEV8 with its alloreactive properties is located beneath the peptide and does not directly contact the T cell receptor (TCR). However, the buried mutation induces local rearrangement of the peptide itself to preserve hydrogen bonding interactions between the peptide and the alpha(1) 77 residue. The COOH terminus of the peptide main chain is tugged toward the alpha(1)-helix such that its presentation to the TCR is altered. These changes increase the stability of the allogeneic peptide-major histocompatibility complex (pMHC) complex and increase complementarity in the TCR-pMHC interface, placing greater emphasis on recognition of the pMHC by the TCR beta-chain, evinced by an increase in shape complementarity, buried surface area, and number of TCR-pMHC contacting residues. A nearly fourfold increase in the number of beta-chain-pMHC contacts is accompanied by a concomitant 64% increase in beta-chain-pMHC shape complementarity. Thus, the allogeneic mutation causes the same peptide to be presented differently, temporally and spatially, by the allogeneic and syngeneic MHCs.


Subject(s)
H-2 Antigens/chemistry , Histocompatibility Antigens Class I/chemistry , Major Histocompatibility Complex , Mutagenesis , Receptors, Antigen, T-Cell/chemistry , Crystallography, X-Ray , Isoantigens/chemistry , Models, Molecular , Peptide Fragments/chemistry , Protein Conformation
11.
J Exp Med ; 182(6): 1739-50, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7500018

ABSTRACT

The identity of allogeneic peptide/major histocompatibility complex (MHC) complexes that elicit vigorous T cell responses has remained an interesting problem for both practical and theoretical reasons. Although a few abundant MHC class I-bound peptides have been purified and sequenced, identifying the unique T cell-stimulating peptides from among the thousands of existing peptides is still a very difficult undertaking. In this report, we identified the antigenic peptide that is recognized by an alloreactive bm1 anti-B6 T cell clone using a novel genetic strategy that is based upon measurement of T cell receptor occupancy in single T cells. Using lacZ-inducible T cells as a probe, we screened a splenic cDNA library in transiently transfected antigen-presenting cells (APCs) and isolated a cDNA clone that allowed expression of the appropriate peptide/Kb MHC complex in APC. The antigenic octapeptide (SVVEFSSL) exactly matched the consensus Kb MHC motif, but was surprisingly encoded by a non-ATG defined translation reading frame. Furthermore, the abundance of the naturally processed analog in untransfected cells was estimated to be <10 copies per cell. These results illustrate a novel strategy for identifying T cell-stimulating antigens in general and directly show that alloreactive T cells can respond to rather rare peptide/MHC complexes. These results also suggest that the total pool of processed peptides expressed on the APC surface may include those generated by cryptic translation of normally expressed transcripts.


Subject(s)
Antigen-Presenting Cells/immunology , H-2 Antigens/immunology , Isoantigens/immunology , Oligopeptides/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , DNA Primers/chemistry , Isoantigens/chemistry , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligopeptides/chemistry , Tubulin/immunology
12.
J Exp Med ; 184(3): 1017-26, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-9064319

ABSTRACT

The 2C T cell is a CD8+, alloreactive T cell, which recognizes cells bearing Ld and Kbm3 class I major histocompatability complex molecules. Here, we characterize an allopeptide, designated dEV-8, that is a ligand in the Kbm3 molecule for the 2C TCR but is not a ligand in the Ld molecule. By biochemical and immunological properties, dEV-8 is distinct from P2Ca, the Ld allopeptide that is also recognized by the 2C TCR. Using the deduced amino acid sequence of dEV-8, we isolate a candidate endogenous source of the peptide. The endogenous protein, MLRQ, contains a peptide sequence identical to dEV-8. This degenerate recognition of two distinct peptide/MHC complexes by a single TCR has important implications for understanding allorecognition.


Subject(s)
Histocompatibility Antigens Class I/immunology , Isoantigens/isolation & purification , Receptors, Antigen, T-Cell/immunology , Amino Acid Sequence , Animals , Base Sequence , Cattle , Chromatography, High Pressure Liquid , Humans , Isoantigens/chemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Mapping , Sequence Homology, Nucleic Acid , Substrate Specificity
13.
Transpl Int ; 23(11): 1154-63, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20536791

ABSTRACT

Memory T cells are known to play a key role in prevention of allograft tolerance in alloantigen-primed mice. Here, we used an adoptively transferred memory T cell model and an alloantigen-primed model to evaluate the abilities of different combinations of monoclonal antibodies (mAb) to block key signaling pathways involved in activation of effector and memory T cells. In the adoptively transferred model, the use of anti-CD134L mAb effectively prevented activation of CD4(+) memory T cells and significantly prolonged islet survival, similar to the action of anti-CD122 mAb to CD8(+) memory T cells. In the alloantigen-primed model, use of anti-CD134L and anti-CD122 mAbs in addition to co-stimulatory blockade with anti-CD154 and anti-LFA-1 prolonged secondary allograft survival and significantly reduced the proportion of memory T cells; meanwhile, this combination therapy increased the proportion of regulatory T cells (Tregs) in the spleen, inhibited lymphocyte infiltration in the graft, and suppressed alloresponse of recipient splenic T cells. However, we also detected high levels of alloantibodies in the serum which caused high levels of damage to the allogeneic spleen cells. Our results suggest that combination of four mAbs can significantly suppress the function of memory T cells and prolong allograft survival in alloantigen primed animals.


Subject(s)
Immunologic Memory , Isoantigens/chemistry , Lymphocyte Activation , Animals , Diabetes Complications/immunology , Female , Interleukin-2 Receptor beta Subunit/biosynthesis , Islets of Langerhans/cytology , Isoantibodies/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Receptors, OX40/biosynthesis , Time Factors , Transplantation Tolerance/immunology , Transplantation, Homologous/methods
14.
Curr Opin Organ Transplant ; 15(4): 512-25, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20616723

ABSTRACT

PURPOSE OF REVIEW: There is ample evidence indicating a pathologic role for minor histocompatibility antigens in inciting graft-versus-host disease in major histocompatibility complex (MHC)-matched bone marrow transplantation and rejection of solid organ allografts. Here we review the current knowledge of the genetic and biochemical bases for the cause of minor histoincompatibility and the structural basis for the recognition of the resulting alloantigens by the T-cell receptor. RECENT FINDINGS: Recent evidence indicates that we as independently conceived individuals are genetically unique, thus, offering a mechanism for minor histoincompatibility between MHC-identical donor-recipient pairs. Furthermore, advances in delineating the mechanisms underlying antigen cross-presentation by MHC class I molecules and a critical role for autophagy in presenting cytoplasmic antigens by MHC class II molecules have been made. These new insights coupled with the X-ray crystallographic solution of several peptide/MHC-T-cell receptor structures have revealed mechanisms of histoincompatibility. SUMMARY: On the basis of these new insights, ways to test for allograft compatibility and concoction of immunotherapies are discussed.


Subject(s)
Graft Rejection/immunology , Graft Survival , Graft vs Host Disease/immunology , Isoantigens/immunology , Minor Histocompatibility Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transplantation Tolerance , Animals , Graft Rejection/prevention & control , Graft vs Host Disease/prevention & control , Humans , Immunosuppression Therapy/methods , Inflammation/immunology , Isoantigens/chemistry , Isoantigens/genetics , Ligands , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/genetics , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Transplantation, Homologous
15.
J Liposome Res ; 18(1): 91-3, 2008.
Article in English | MEDLINE | ID: mdl-18348075

ABSTRACT

How do tissues hide themselves from the body's natural defense system? Anonymity is difficult to sustain in the blood stream. Almost all surfaces become tagged by polyionic molecules, such as proteins, within milliseconds of being exposed to plasma (which, after all, is an abundant source of animal glue). Both the tagging and the gluing are spontaneous events and result from the release of electrostatically bound water when ionic groups of the surface and the protein mutually satisfy their respective charges. The driving forces responsible for the spontaneity of the adsorption/gluing process are various attractive forces and the disordering of bound water leading to a gain in entropy. But suppose the surface of an intruder had no surface charges, causing it to resemble the surface of a slice of (bulk) water. The polyionic proteins would not find anything to tag, there is no entropy gain, there are no attractive interactions, and the interloper would be invisible.


Subject(s)
Immune System , Liposomes/chemistry , Animals , Drug Delivery Systems , Female , Humans , Hydrogen-Ion Concentration , Hydrolysis , Isoantigens/chemistry , Lecithins/chemistry , Lysophospholipase/chemistry , Major Histocompatibility Complex , Mice , Pregnancy
16.
Front Immunol ; 9: 2284, 2018.
Article in English | MEDLINE | ID: mdl-30364159

ABSTRACT

Alloreactivity compromising clinical outcomes in stem cell transplantation is observed despite HLA matching of donors and recipients. This has its origin in the variation between the exomes of the two, which provides the basis for minor histocompatibility antigens (mHA). The mHA presented on the HLA class I and II molecules and the ensuing T cell response to these antigens results in graft vs. host disease. In this paper, results of a whole exome sequencing study are presented, with resulting alloreactive polymorphic peptides and their HLA class I and HLA class II (DRB1) binding affinity quantified. Large libraries of potentially alloreactive recipient peptides binding both sets of molecules were identified, with HLA-DRB1 generally presenting a greater number of peptides. These results are used to develop a quantitative framework to understand the immunobiology of transplantation. A tensor-based approach is used to derive the equations needed to determine the alloreactive donor T cell response from the mHA-HLA binding affinity and protein expression data. This approach may be used in future studies to simulate the magnitude of expected donor T cell response and determine the risk for alloreactive complications in HLA matched or mismatched hematopoietic cell and solid organ transplantation.


Subject(s)
Antigens/immunology , Stem Cell Transplantation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Algorithms , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Graft vs Host Disease/etiology , HLA Antigens/immunology , Histocompatibility/genetics , Histocompatibility/immunology , Humans , Isoantigens/chemistry , Isoantigens/immunology , Isoantigens/metabolism , Models, Theoretical , Peptides/immunology , Peptides/metabolism , Protein Binding , Stem Cell Transplantation/adverse effects , Tissue Donors
17.
Immunohematology ; 23(4): 146-9, 2007.
Article in English | MEDLINE | ID: mdl-18284304

ABSTRACT

The low-prevalence MNS blood group antigenTSEN is located at the junction of glycophorin A (GPA) to glycophorin B (GPB) in several hybrid glycophorin molecules. Extremely rare people have RBCs with a double dose of the TSEN antigen and have made an antibody to a high-prevalence MNS antigen. We report the first patient who is heterozygous for GYP.JL and Mk. During prenatal tests,an alloantibody to a high-prevalence antigen was detected in the serum of a 21-year-old Hispanic woman. The antibody detected an antigen resistant to treatment by papain, trypsin, alpha-chymotrypsin, or DTT. The antibody was strongly reactive by the IAT with all RBCs tested except those having the MkMk, GP.Hil/GP.Hil, or GP.JL/GP.JL phenotypes. The patient's RBCs typed M+N-S+/-s-U+, En(a+/-), Hut-, Mi(a-), Mur-, Vw-, Wr(a-b-), and were TSEN+, MINY+. Reactivity with Glycine soja suggested that her RBCs had a decreased level of sialic acid. Immunoblotting showed the presence of monomer and dimer forms of a GP(A-B) hybrid and an absence of GPA and GPB. Sequencing of DNA and PCR-RFLP using the restriction enzyme RsaI confirmed the presence of a hybrid GYP(AB). The patient's antibody was determined to be anti-EnaFR. She is the first person reported with the GP.JL phenotype associated with a deletion of GYPA and GYPB in trans to GYP.JL.


Subject(s)
Glycophorins/chemistry , Glycophorins/immunology , Isoantibodies/blood , Isoantigens/chemistry , MNSs Blood-Group System/genetics , Phenotype , Adult , Base Sequence , Blood Grouping and Crossmatching/methods , Erythrocyte Membrane/chemistry , Erythrocyte Membrane/immunology , Erythrocytes/chemistry , Erythrocytes/immunology , Female , Genotype , Hemagglutination/immunology , Humans , Infant, Newborn , Isoantibodies/immunology , Polymorphism, Restriction Fragment Length/immunology , Pregnancy , Spain
18.
Proteins ; 63(3): 512-26, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16470819

ABSTRACT

Since determining the crystallographic structure of all peptide-MHC complexes is infeasible, an accurate prediction of the conformation is a critical computational problem. These models can be useful for determining binding energetics, predicting the structures of specific ternary complexes with T-cell receptors, and designing new molecules interacting with these complexes. The main difficulties are (1) adequate sampling of the large number of conformational degrees of freedom for the flexible peptide, (2) predicting subtle changes in the MHC interface geometry upon binding, and (3) building models for numerous MHC allotypes without known structures. Whereas previous studies have approached the sampling problem by dividing the conformational variables into different sets and predicting them separately, we have refined the Biased-Probability Monte Carlo docking protocol in internal coordinates to optimize a physical energy function for all peptide variables simultaneously. We also imitated the induced fit by docking into a more permissive smooth grid representation of the MHC followed by refinement and reranking using an all-atom MHC model. Our method was tested by a comparison of the results of cross-docking 14 peptides into HLA-A*0201 and 9 peptides into H-2K(b) as well as docking peptides into homology models for five different HLA allotypes with a comprehensive set of experimental structures. The surprisingly accurate prediction (0.75 A backbone RMSD) for cross-docking of a highly flexible decapeptide, dissimilar to the original bound peptide, as well as docking predictions using homology models for two allotypes with low average backbone RMSDs of less than 1.0 A illustrate the method's effectiveness. Finally, energy terms calculated using the predicted structures were combined with supervised learning on a large data set to classify peptides as either HLA-A*0201 binders or nonbinders. In contrast with sequence-based prediction methods, this model was also able to predict the binding affinity for peptides to a different MHC allotype (H-2K(b)), not used for training, with comparable prediction accuracy.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Isoantigens/metabolism , Models, Molecular , Peptide Fragments/metabolism , Binding Sites , Histocompatibility Antigens Class I/chemistry , Isoantigens/chemistry , Peptide Fragments/chemistry , Predictive Value of Tests , Protein Binding , Protein Conformation , Protein Structure, Secondary
19.
Transplantation ; 82(8): 1085-92, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-17060858

ABSTRACT

BACKGROUND: Blockade of T cell costimulation by treatment with donor-specific transfusion (DST) and anti-CD154 monoclonal antibody (mAb) induces prolonged allograft survival in mice. This effect is due in part to deletion of host CD8 and CD4 T cells that recognize alloantigen by direct presentation. The fate of host CD4 T cells that recognize alloantigen by indirect presentation, however, is unclear. METHODS: We studied Tg361 TCR transgenic CD4 T cells that recognize alloantigen by indirect presentation. Carboxyfluorescein diacetate, succinimidyl ester-labeled Tg361 cells were adoptively transferred into syngeneic nontransgenic recipients and their fate in the peripheral blood, spleen, and lymph nodes following treatment with DST and anti-CD154 was analyzed. RESULTS: Treatment of mice with DST plus anti-CD154 mAb does not delete Tg361 CD4 T cells, but instead renders them hyporesponsive to rechallenge with alloantigen. Mice circulating hyporesponsive CD4 T cells also fail to reject skin allografts. The hyporesponsive state of the T cells is not reversed by the addition of interleukin-2, anti-CD28 mAb, or an agonistic anti-CD134 mAb in the presence of antigen. These T cells are capable of activation, however, as evidenced by in vitro proliferation in response to anti-CD3 mAb. CONCLUSIONS: These results demonstrate that costimulation blockade can induce hyporesponsiveness of host CD4 T cells recognizing alloantigens by indirect presentation, thus prolonging graft survival by a mechanism that does not involve deletion of alloreactive T cells.


Subject(s)
Antigen Presentation , Isoantigens/chemistry , T-Lymphocytes/cytology , Animals , CD4-Positive T-Lymphocytes/cytology , CD40 Ligand/biosynthesis , CD8-Positive T-Lymphocytes/cytology , Cricetinae , Fluoresceins/pharmacology , Graft Survival , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Rats
20.
Med Oncol ; 23(2): 273-82, 2006.
Article in English | MEDLINE | ID: mdl-16720928

ABSTRACT

A variety of approaches have been used to deliver tumor-associated antigens (TAA) in conjunction with dendritic cells (DC) as cellular adjuvants. DC derived from monocytic precursors have been pulsed with whole tumor antigen using a variety of strategies and have been demonstrated to induce CD4+ and CD8+ antitumor responses. In the present study, monocyte-derived DC have been pulsed with lysate from an allogeneic melanoma cell line, A-375, and used to repeatedly stimulate T cells. The resultant T cells were examined for cytotoxic activity against A-375 targets as well as the HLA A2-positive melanoma cell line DFW. Uptake of FITC-labeled melanoma lysate by DC established that lysate of melanoma cells was efficiently endocytosed. Stimulation with lysate-pulsed DC resulted in strong proliferative responses by T cells, which could be inhibited by antibodies against both MHC class I and class II. T cells stimulated in vitro with lysate-pulsed DC demonstrated potent cytotoxicity against the melanoma targets which were blocked by antibodies against MHC class I. Lysate-pulsed DC also elicited IFN-gamma secretion by T cells as measured in an ELISPOT assay. We have also examined the ability of lysate-pulsed DC to present melanoma-associated antigens to T cells. ELISPOT assays with synthetic peptides of melanoma-associated antigens, such as gp100, mage1, NY-ESO, and MART-1, revealed that lysate-pulsed DC could stimulate T cells in an antigen-specific manner. The results demonstrate that lysate from allogeneic tumor cells may be used as a source of antigens to stimulate tumor-specific T cells in melanoma.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Melanoma/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/pharmacology , Cell Line, Tumor , Coculture Techniques , Humans , Isoantigens/chemistry , Isoantigens/immunology , Isoantigens/pharmacology , K562 Cells , Melanoma/chemistry , Monocytes
SELECTION OF CITATIONS
SEARCH DETAIL